Although mantle cell lymphoma (MCL) is a rare subtype of non–Hodgkin lymphoma, proactive research efforts fueled by challenges in the management of MCL have led to an increase in median overall survival (OS) of 2.5 years in the mid 1990s to beyond 5 years nowadays. This improvement is due mostly to the use of dose-intensive strategies, particularly cytarabine-containing regimens [with or without high-dose therapy (HDT) followed by autologous stem cell transplantation (ASCT) consolidation], which are associated with deeper remission (and higher molecular complete response rate), as well as better salvage therapies. Along this line, MCL became the first lymphoma for which four novel agents have been approved in the relapsed/refractory setting: temsirolimus, lenalidomide, ibrutinib, and bortezomib (the last agent approved both in relapsed/refractory disease and in first-line combination therapy). In addition, the use of rituximab maintenance has helped reduce relapse rates and improve outcome. However, in routine practice (i.e., outside clinical trials), the outcome of MCL remains overall unchanged with standard immunochemotherapy, and even after HDT-ASCT, most patients still relapse and frequently develop chemoresistance. The persistent lack of consensus for the treatment of MCL explains the rather impressive variability in management of these patients. The integration of newer therapies, either in combination with immunochemotherapy or as consolidation/maintenance postinduction, offers new opportunities for patients with MCL. This review highlights how such developments can help refine the current MCL paradigm. Clin Cancer Res; 21(17); 3853–61. ©2015 AACR.

Mantle cell lymphoma (MCL) accounts for about 6% of all non–Hodgkin lymphomas (NHL), although recent reports suggest an increase in incidence (1). Median age at diagnosis is mid-60s, with a 3:1 male:female ratio and frequent extranodal involvement (bone marrow, blood, and gastrointestinal tract particularly; refs. 2, 3). As WHO first recognized MCL as a separate entity in 1994, the median overall survival (OS) has improved from <2.5 to >5 years, although recent reports suggest that for the overall population (i.e., outside clinical trials), median OS remains <3 years (4).

Derived from mostly antigen-naïve cells, MCL cells proliferate in the mantle zone around germinal centers (2), with morphologic (diffuse, nodular, mantle zone) as well as cytologic variants (small cells, pleomorphic, blastoid). Diagnosis suspected on immunophenotype (CD20+, CD5+, CD23, and FMC7+) requires confirmation by CYCLIN D1 overexpression due to t(11;14) translocation, which is generally identified by FISH rather than cytogenetics (2, 3, 5). Rare cases of CYCLIN D1–negative MCL show CYCLIN D2 or D3 overexpression (through rearrangements with IGH or IGL loci) and share similar clinical behavior and outcome with CYCLIN D1–positive cases (6).

Outcome in MCL may be predicted by the MCL International Prognostic Index (MIPI) based on age, Eastern Cooperative Oncology Group (ECOG) performance status, lactate dehydrogenase (LDH) levels, and white blood cell counts (7–10). MIPI stratifies patients into 3 risk groups, low, intermediate, and high, with median OS not reached, 51 months, and 29 months, respectively (7). Gene expression profiling (GEP) revealed a proliferation signature with dramatic impact on outcome (median OS ranging from <1 to 7+ years; ref. 6). Ki-67 (or MIB1) has been used as a surrogate marker of proliferation (</≥30%), showing value independently of MIPI (11). However, Ki-67 evaluation by immunohistochemistry remains by nature semiquantitative and is found in most cases at baseline in the <10% to <30% range. Rare cases presenting with high Ki-67 at baseline, often carrying DEL17P/P53 mutation; both features are consistent with blastoid variant and/or are more commonly seen with successive relapses (i.e., clonal evolution). Other prognostic variables have been reported in MCL, including β2M, miRNAs, and complex karyotype, whereas overall P53 abnormalities are likely underestimated (2, 12). SOX11 expression can help diagnose CYCLIN D1–negative cases and seems to correlate with outcome (13), although consensus on a validated analysis tool for SOX11 remains to be determined (14). Comprehensive genomic studies have confirmed the complex mutational landscape of MCL, which contributes to pathogenesis and disease progression, including mutations in ATM, P53, BIRC3, WHSC1, and NOTCH1 (15).

About 10% to 15% of patients with MCL presenting with high leukemic phase, minimal, or no lymphadenopathy, and/or large splenomegaly are SOX11-negative and show distinct GEP, carry a more indolent course, and should be managed conservatively (1, 6, 16–18). Genetically, these patients with “truly indolent MCL” typically show somatic mutations, which like in chronic lymphocytic leukemia correlates with higher genetic instability (fewer secondary genetic abnormalities over time than classic MCL), although some cases may acquire DEL17P/P53 mutation (1, 2, 19). In situ MCL lesions have been reported (usually an incidental finding) with a very indolent behavior, distinguishable from mantle zone variant or overt MCL and should be monitored (20). More challenging are low-risk patients with MCL (low tumor burden, low Ki-67) who can also be monitored (16), as illustrated in the watch-and-wait group from the recent Nordic Lymphoma Group observational report for ≥2 years of untreated patients who had a 3-year OS rate of 79% (9). Putting aside the caveats related to these retrospective series, up to one third of MCL at diagnosis can be monitored with a time to therapy of 1 year, although there are no clear criteria to identify such cases.

Most patients present with “classic” MCL and are considered for treatment at diagnosis (21, 22). Selection of induction therapy is usually based on patient's ability to tolerate intensive therapy, thereby involving age (</≥65 years) and comorbidities. If feasible, there is clear consensus that intensive strategies lead to significant improvement in progression-free survival (PFS; >5 years) over standard immunochemotherapy such as R-CHOP, which has been proven both in clinical trials (23) and routine practice (24). In the MCL NCCN database, 3-year PFS rate for R-CHOP was 18% (3 times lower than with dose-intensive strategies: 55%–58%), and no OS benefit was shown for pooled dose-intense combinations versus R-CHOP (24). The benefit of cytarabine-containing induction was suggested in several phase II studies; R-CHOP/R-DHAP (3 cycles of each) in a series of 60 patients (aged ≤66 years) followed by autologous stem cell transplantation (ASCT) led to significant improvement in complete responses (CR; 57% vs. 15%) over historical R-CHOP alone, translating into an impressive median event-free survival (EFS) of 83.9 months, median PFS of 84 months, and 5-year OS rate of 75% (median not reached; ref. 25). Several other regimens corroborated those findings, including R-hyper-CVAD (26, 27), NORDIC MCL2 (28, 29), CALGB regimens (30), among others (Table 1; refs. 25–31). The benefit of regimens containing HD-AraC was confirmed in a large (>200 patients/arm) randomized European MCL Network trial comparing arm A [R-CHOP→myeloablative radiochemotherapy (TBI, cyclophosphamide) and ASCT] versus arm B [alternating R-CHOP/R-DHAP→HD-Ara-C–containing myeloablative regimen (TBI, Ara-C, melphalan) and ASCT; abstract-only references (32, 33)]. At a median follow-up of 51 months, results showed superiority for the HD-AraC arm for time-to-treatment failure (primary endpoint: 46 vs. 88 months, P = 0.038), remission duration (49 vs. 84 months, P = 0.0001), and OS (82 months vs. not reached, P = 0.045). Although the proportion of patients receiving ASCT was similar (72% vs. 73%), benefit in the HD-AraC arm was attributed to higher and earlier CR rate paralleled by increased molecular CR, translating into fewer relapses in responders (n = 81 vs. n = 40; ref. 34).

Table 1.

Clinical efficacy of select induction regimens in previously untreated MCL

Treatment/studyORRCREfficacy endpointsOS
Aggressive treatment: younger and transplant-eligible patients 
 R-hyper-CVAD/R-MA (n = 97; refs. 26, 27) 97% 87% Median 10-year TTF = 4.6 years (5.9 years for ≤65 years) 3-year OS rate = 82% (74% for ≥65 years) 
    Median 10-year OS = NR 
 NORDIC MCL2: maxi-CHOP alternating with rituximab + HD-AraC (n = 160; ref. 28) 96% 54% 6-year EFSa rate = 56% 6-year OS rate = 70% 
   6-year PFS rate = 66%  
 Sequential R-CHOP/R-DHAP (n = 60; ref. 25) 87% 57% Median DFS = 78 mo Median OS = NR 
   Median EFSb = 83.9 mo 5-year OS rate = 75% 
   5-year EFS rate = 64%  
   Median PFS = 84 mo  
 CALGB immunochemotherapy regimen (n = 78; ref. 30) 88% 69% 2-year PFS rate = 76% 2-year OS rate = 87% 
   5-year PFS rate = 56% 5-year OS rate = 64% 
Less aggressive: older (aged >65 years) and/or transplant-ineligible patients 
 R-CHOP (n = 239) vs. R-FC (n = 246; ref. 39) 86% vs. 78% 34% vs. 40% Median TTF = 28 vs. 26 mo 4-year OS rate = 62% vs. 47% 
   Median DOR = 36 vs. 37 mo R-CHOP + R = 87% 
    R-CHOP + IFNα = 63% 
 R-CHOP (n = 62) vs. CHOP (n = 59; ref. 38) 94% vs. 75% (P = 0.0054) 34% vs. 7% (P = 0.00024) Median TTF = 21 vs. 14 mo (P = 0.0131) 
   No difference in PFS or OS 
   2-year survival rate: 77% for both arms 
 R-CHOP/R-AraC + R-F/AraC + CHOP + R maintenance in responders (n = 60; ref. 40) 95% 87% 4-year PFS rate = 70% 4-year OS rate = 72% 
   4-year EFSa rate = 66%  
 ECOG E1405: VcR-CVAD + R maintenance or ASCT (n = 75; ref. 46) 95% 68% 3-year PFS rate = 72% 3-year OS rate = 88% 
 BR (n = 261) vs. R-CHOP (n = 253; ref. 47) 93% vs. 91% 40% vs. 30% (P = 0.021) Median PFS rate = 69.5 vs. 31.2 mo (P < 0.0001) No difference in OS 
 BR [n = 213 (n = 36 MCL)] vs. R-CHOP/R-CVP [n = 206 (n = 38 MCL); BRIGHT; ref. 48) 94% vs. 84% 51% vs. 24% (P = 0.0180) — — 
 R-BAC (n = 20 first-line; n = 20 R/R; ref. 51) 100% first-line 95% first-line 2-year PFS rate = 95% first-line — 
 80% R/R 74% R/R 2-year PFS rate = 70% R/R  
Treatment/studyORRCREfficacy endpointsOS
Aggressive treatment: younger and transplant-eligible patients 
 R-hyper-CVAD/R-MA (n = 97; refs. 26, 27) 97% 87% Median 10-year TTF = 4.6 years (5.9 years for ≤65 years) 3-year OS rate = 82% (74% for ≥65 years) 
    Median 10-year OS = NR 
 NORDIC MCL2: maxi-CHOP alternating with rituximab + HD-AraC (n = 160; ref. 28) 96% 54% 6-year EFSa rate = 56% 6-year OS rate = 70% 
   6-year PFS rate = 66%  
 Sequential R-CHOP/R-DHAP (n = 60; ref. 25) 87% 57% Median DFS = 78 mo Median OS = NR 
   Median EFSb = 83.9 mo 5-year OS rate = 75% 
   5-year EFS rate = 64%  
   Median PFS = 84 mo  
 CALGB immunochemotherapy regimen (n = 78; ref. 30) 88% 69% 2-year PFS rate = 76% 2-year OS rate = 87% 
   5-year PFS rate = 56% 5-year OS rate = 64% 
Less aggressive: older (aged >65 years) and/or transplant-ineligible patients 
 R-CHOP (n = 239) vs. R-FC (n = 246; ref. 39) 86% vs. 78% 34% vs. 40% Median TTF = 28 vs. 26 mo 4-year OS rate = 62% vs. 47% 
   Median DOR = 36 vs. 37 mo R-CHOP + R = 87% 
    R-CHOP + IFNα = 63% 
 R-CHOP (n = 62) vs. CHOP (n = 59; ref. 38) 94% vs. 75% (P = 0.0054) 34% vs. 7% (P = 0.00024) Median TTF = 21 vs. 14 mo (P = 0.0131) 
   No difference in PFS or OS 
   2-year survival rate: 77% for both arms 
 R-CHOP/R-AraC + R-F/AraC + CHOP + R maintenance in responders (n = 60; ref. 40) 95% 87% 4-year PFS rate = 70% 4-year OS rate = 72% 
   4-year EFSa rate = 66%  
 ECOG E1405: VcR-CVAD + R maintenance or ASCT (n = 75; ref. 46) 95% 68% 3-year PFS rate = 72% 3-year OS rate = 88% 
 BR (n = 261) vs. R-CHOP (n = 253; ref. 47) 93% vs. 91% 40% vs. 30% (P = 0.021) Median PFS rate = 69.5 vs. 31.2 mo (P < 0.0001) No difference in OS 
 BR [n = 213 (n = 36 MCL)] vs. R-CHOP/R-CVP [n = 206 (n = 38 MCL); BRIGHT; ref. 48) 94% vs. 84% 51% vs. 24% (P = 0.0180) — — 
 R-BAC (n = 20 first-line; n = 20 R/R; ref. 51) 100% first-line 95% first-line 2-year PFS rate = 95% first-line — 
 80% R/R 74% R/R 2-year PFS rate = 70% R/R  

Abbreviations: BAC, bendamustine, cytarabine; CALGB, Cancer and Leukemia Group B; CHOP, cyclophosphamide, doxorubicin, vincristine, prednisone; CVAD, cyclophosphamide, vincristine, doxorubicin, dexamethasone; CVP, cyclophosphamide, vincristine, prednisolone; DHAP, dexamethasone, AraC, cisplatin; FC, fludarabine, cyclophosphamide; MA, methotrexate, cytarabine; MR, maintenance rituximab; NR, not reached; R, rituximab; R/R, relapsed/refractory; TTF, time to treatment failure; VcR-CVAD, bortezomib, rituximab, modified hyper-CVAD [hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone without alternating cycles of cytarabine (AraC) and methotrexate].

aEFS was calculated from study entry to treatment failure due to progression, toxicity, or death from any cause.

bEFS was defined as relapse after CR/CRu.

In summary, MCL in young and/or fit patients should be managed using a cytarabine-containing intensive approach with or without ASCT (27, 35), which translate into PFS in excess of 5 years. However, late relapses still do occur (28, 29), supporting emerging strategies of maintenance and/or consolidation particularly with biologic agents (including rituximab), as shown recently after R-DHAP-ACST, where maintenance post-ASCT showed significantly improved EFS and PFS (36). Ongoing trials evaluating the role of other biologicals, particularly ibrutinib, after induction therapy will help refine the best approach to prevent recurrence and will likely be based on minimal residual disease (MRD) status.

R-CHOP–like induction

For older patients or those deemed ineligible for dose-intensive therapy, R-CHOP has been the default backbone. With an about 80% overall response rate (ORR), CR remains low (30%–40%) and most patients relapse as shown by the 18% to 25% 2-year PFS (Table 1; refs. 37–39). Building upon R-CHOP, several regimens have explored strategies incorporating newer therapies such as fludarabine (40) or HD-cytarabine (ongoing elderly trial in EU), replacement of cisplatinum compounds with other derivatives (e.g., oxaliplatinum), and addition of novel biologicals such as bortezomib—promising in early phase II study (41)—leading to establishment of a new standard in frontline MCL as part of the LYM-3002 trial (42). The pivotal LYM-3002 study comparing VcR-CAP with R-CHOP showed significantly higher CR rate and improved median PFS in the bortezomib-containing arm (24.7 vs. 14.4 months, P < 0.001, respectively; ref. 42), leading to extended FDA approval for bortezomib in previously untreated MCL in October 2014 (43). Because of hyper-CVAD–related toxicity (44), a modified combination (no cytarabine or methotrexate) was developed with results intermediate between R-CHOP and standard hyper-CVAD, but better tolerated, also offering a platform for bortezomib combinations (45). This benefit was confirmed in the multicenter setting by the ECOG E1405 study, in which VcR-CVAD followed by maintenance rituximab in newly diagnosed MCL showed an ORR of 95%, with a 68% CR/CR unconfirmed (CRu), and 3-year PFS of 72% (46).

Bendamustine-based induction

The bendamustine + rituximab (BR) regimen was established by the StiL trial, comparing BR (n = 261) versus R-CHOP (n = 253) in indolent NHL (iNHL, >50% follicular lymphoma; ref. 47). BR showed superior CR (40% vs. 30% R-CHOP, P = 0.021) and median PFS (69.5 vs. 31.2 months, P < 0.0001) for all patients, whereas in the subset of patients with MCL (n = 46 BR and n = 48 R-CHOP), PFS was 35.4 for the BR arm versus 22.1 months for R-CHOP (P = 0.0044, Table 1). In an effort to replicate these data, the U.S.-based BRIGHT trial in iNHL and MCL compared BR [n = 213 (n = 36 MCL)] with R-CHOP/R-CVP [n = 206 (n = 38 MCL)] suggesting a higher CR in favor of BR (31% vs. 25%), although some patients received only R-CVP (48). BR toxicity was less favorable than in the StiL trial, and PFS was not clearly different (47, 48). Despite of the lack of difference in OS, the overall favorable short-term toxicity profile, particularly in the StiL trial, established a new standard in MCL, offering a new backbone for combinations such as BR + bortezomib, which showed very promising results in phase II studies in relapsed/refractory MCL (83% ORR, 52% CR; ref. 49). The LYSA group recently presented impressive results with frontline RiBVD (rituximab, bendamustine, bortezomib, and dexamethasone) in 74 patients with MCL (no maintenance) with a 74% CR/CRu alongside measurable molecular remission in blood (83%) and bone marrow (74%), translating into an estimated 24-month PFS and OS rates of 69% and 80%, respectively (50). Other studies combining BR with ibrutinib (SHINE trial) or lenalidomide are ongoing. The addition of cytarabine to BR (R-BAC) was reported in patients with MCL aged ≥65 years; in this small series, results were impressive, with 100% ORR (95% CR) and 95% 2-year PFS rate for previously untreated MCL (n = 20), and 80% ORR (74% CR) with a 70% 2-year PFS rate for relapsed/refractory MCL (n = 20; ref. 51).

Bortezomib was the first novel agent integrated into upfront MCL strategies, either with standard therapy as seen above with R-CHOP (SWOG-S0601 study abstract; refs. 52, 53) or after high-dose therapy (HDT) and ASCT (54). Lenalidomide, with durable responses in the relapsed/refractory setting (55), was combined with rituximab in relapsed/refractory patients with promising results (56). A similar combination in the frontline setting (12 cycles R2 induction followed by R2 maintenance until progression) showed rapid responses (median time to response, 2.8 months), leading to an ORR of 84%, including 53% CR/CRu and an 84% estimated 2-year PFS (57). A number of ongoing studies are looking at other biologic combinations, for example, BR ± ibrutinib (NCT01776840) or BR ± bortezomib followed by rituximab ± lenalidomide maintenance in elderly patients (ECOG1411, NCT01415752). Both lenalidomide and ibrutinib are also being evaluated as part of HDT–ASCT approaches in combination or sequentially as maintenance.

The European MCL Network Clinical Intergroup phase III study in patients aged ≥60 years (median, 70 years) evaluated R-CHOP21 (8 cycles) versus R-FC (6 cycles) followed by a second randomization in responders for maintenance with rituximab versus IFNα (39). Although similar ORR and CR were seen with R-CHOP or R-FC induction, the 4-year OS rates differed significantly (62% vs. 47%, P = 0.0005), and patients experienced higher mortality and toxicity with R-FC induction. In addition, in the R-CHOP arm, rituximab maintenance significantly reduced relapse and improved OS at 4 years (87% vs. 63%, P = 0.0005). Of notice, by design, patients remained on rituximab maintenance until progression (some still on study beyond 5 years). As expected, hematologic toxicity was more common with R-FC than R-CHOP; in the maintenance arms, toxicity from IFNα (vs. rituximab) led to lower compliance.

In the NORDIC MCL2 trial, rituximab was added as “preemptive maintenance” based on MRD monitoring with PCR conversion taken as an indicator of molecular relapse (i.e., not considered failures), leading most patients to convert again to PCR negativity after additional rituximab (29). Several ongoing trials are investigating the potential advantage of lenalidomide ± rituximab maintenance following R-chemotherapy (NCT01035463, NCT01865110) or lenalidomide/rituximab (NCT01996865). Bortezomib has also been tested as an optional agent for maintenance in 65 patients receiving bortezomib maintenance after R-CHOP SWOG-S0601), leading to an estimated doubled 2-year PFS (62% bortezomib vs. 30% historical R-CHOP alone; ref. 52). Given the radiosensitivity of iNHL and MCL, radioimmunotherapy was tested as consolidation after 4 cycles of R-CHOP (n = 56) showing promising results, with an ORR of 82% (55% CR/CRu), including a tripling of CR rate post-RIT (from 18% to 55%), as well as favorable PFS compared with historical controls (58). On the other hand, in the NORDIC MCL3 trial, the addition of RIT consolidation [ibritumomab tiuxetan (0.4 mCi/kg)] pre-HDT (if in CRu/PR) showed no clear benefit (59). While still being evaluated in numerous ongoing trials, the use of postinduction strategies as maintenance/consolidation, even following HDT-ASCT, is now supported by randomized trials showing the benefit of rituximab in both younger (36) and elderly (39) patients with MCL. The next step will be to build on maintenance rituximab by using biologic combinations, although such strategies should be targeted to patients at higher risk of recurrence because of added potential toxicities and cost issues.

Although high CR rates can be obtained with either dose-intensive strategies or combinations of biologicals with standard therapy, a significant proportion of patients still relapse. Two strategies have been evaluated to appreciate the quality of response in MCL: functional imaging [positron emission tomography (PET)] and MRD assessment. Retrospective studies of PET following R-hyper-CVAD induction showed a significant association between posttreatment PET positivity and outcome, whereas interim PET was not predictive (60). PET negativity before upfront ASCT was associated with superior PFS and OS in MCL (61) but not if induction was R-CHOP only (ref. 62; likely because response was not deep enough with R-CHOP). No correlation was found between Ki-67 or MRD measurements by PCR and PET, although patients with PR but negative MRD status seem to do as well as CR patients (34). As in other subtypes of NHL, issues with residual activity and interpretation of SUV values changes (and often-small patient series) limit definite conclusions and recommendations for use in practice, until results of larger prospective studies become available.

The role of MRD status in MCL, although not yet used in routine practice, is gaining interest. Several studies have confirmed, not surprisingly, that the achievement of molecular remission (MR; i.e., molecular CR) in peripheral blood and/or bone marrow following induction immunochemotherapy is significantly associated with a better outcome, independently of other prognostic variables in both elderly and younger patients (34, 63), and should become the goal in upcoming or ongoing immunochemotherapy trials (64).

Multiple chemotherapy- and molecular-based treatments have been used in relapsed/refractory MCL, with no clear standard of care established to date (21, 22). Induction strategies may be carried over into the relapsed setting in an attempt to achieve better response with a different treatment than was initially received. For example, BR showed 75% ORR (50% CR) and median PFS of 18 months in 16 patients with MCL (1–3 prior therapies) and may be considered second-line after non-BR regimens (65). As noted above, R-BAC showed promising results following ≥1 prior treatment (80% ORR, 70% CR), with a 2-year PFS rate of 70% (51). The most common grade 3/4 toxicity associated with R-BAC was transient and reversible myelosuppression (83% grade 3/4 thrombocytopenia).

Until 2013, bortezomib was the only FDA-approved agent for relapsed/refractory MCL in the United States (66, 67), as was temsirolimus in the European Union (68). Two additional newer agents, lenalidomide and ibrutinib, were recently approved in the United States for relapsed/refractory MCL (55, 69). Multiple phase II studies of lenalidomide, an immunomodulator also with direct antineoplastic effects, have provided consistent ORR (28%–53%) with durable activity [median duration of response (DOR), 13.7–16.6 months] in heavily pretreated patients, including those failing bortezomib (NHL-002, NHL-003, and MCL-001 studies; refs. 55, 70, 71). Recently reported results from MCL-002, a randomized phase II study of relapsed/refractory patients with MCL demonstrated superior median PFS following lenalidomide (8.7 months) versus investigator's choice monotherapy (5.2 months; P = 0.004; ref. 72). Also approved in 2013 was ibrutinib, a small-molecule inhibitor of Bruton tyrosine kinase, based on its phase II activity (68% ORR and median DOR 17.5 months) and favorable safety profile (69). Table 2 summarizes key clinical study findings for all 4 approved agents in relapsed/refractory MCL, with lenalidomide and ibrutinib showing durable responses, including following multiple prior therapies (55, 66–71).

Table 2.

Clinical efficacy for approved treatments with relapsed/refractory MCL

PatientsORRCRMedian DOR, moMedian PFS, moMedian OS, mo
Bortezomib (PINNACLE; n = 155; refs. 66, 67) 32% 8% 9.2 6.5 23.5 
Temsirolimus (175/75 mg; n = 54) vs. temsirolimus (175/25 mg; n = 54) vs. investigator's choice (n = 54; ref. 68) 22% vs. 6% vs. 2% 2% vs. 0% vs. 2% 7.1 vs. 3.6 vs. N/A 4.8 vs. 3.4 vs. 1.9 12.8 vs. 10.0 vs. 9.7 
Lenalidomide 
 NHL-002 (n = 15; ref. 70) 53% 20% 13.7 5.6 NR 
 NHL-003 (n = 57; ref. 71) 35% 12% 16.3 8.8 NR 
 MCL-001 (EMERGE; n = 134; ref. 55) 28% 8% 16.6 4.0 19.0 
 MCL-002: Lenalidomide (n = 170) vs. 40% 5% 16.1 8.7 27.9 
Investigator's choice (n = 84; ref. 72) 11% 10.4 5.2 21.2 
Ibrutinib (n = 111; ref. 69) 68% 21% 17.5 13.9 NR 18-mo OS rate = 58% 
PatientsORRCRMedian DOR, moMedian PFS, moMedian OS, mo
Bortezomib (PINNACLE; n = 155; refs. 66, 67) 32% 8% 9.2 6.5 23.5 
Temsirolimus (175/75 mg; n = 54) vs. temsirolimus (175/25 mg; n = 54) vs. investigator's choice (n = 54; ref. 68) 22% vs. 6% vs. 2% 2% vs. 0% vs. 2% 7.1 vs. 3.6 vs. N/A 4.8 vs. 3.4 vs. 1.9 12.8 vs. 10.0 vs. 9.7 
Lenalidomide 
 NHL-002 (n = 15; ref. 70) 53% 20% 13.7 5.6 NR 
 NHL-003 (n = 57; ref. 71) 35% 12% 16.3 8.8 NR 
 MCL-001 (EMERGE; n = 134; ref. 55) 28% 8% 16.6 4.0 19.0 
 MCL-002: Lenalidomide (n = 170) vs. 40% 5% 16.1 8.7 27.9 
Investigator's choice (n = 84; ref. 72) 11% 10.4 5.2 21.2 
Ibrutinib (n = 111; ref. 69) 68% 21% 17.5 13.9 NR 18-mo OS rate = 58% 

Abbreviations: N/A, not available; NR, not reached.

Although overall studies do not support the use of ASCT in the relapsed/refractory setting (73, 74), allogeneic transplantation can be potentially curative (75, 76), although associated with a >50% risk of chronic GVHD. Approaches using haplotransplantation or TH2 amplification (to reduce GVH) or autologous chimeric antigen receptor (CAR) T cells might help implement cell therapy in an often elderly population with chemoresistant disease (77).

The availability of newer agents with efficacy in disease refractory to standard therapies offers new opportunities for risk-adapted and molecular-based targeted therapy, appealing options in an older population. On the basis of early activity of rituximab combined with thalidomide in relapsed/refractory MCL and the improved safety profile with lenalidomide over thalidomide (78), studies combining rituximab with lenalidomide (i.e., R2) show impressive activity compared with either agent alone in the relapsed/refractory (57% ORR, 36% CR and median DOR of 18.9 months; ref. 56) and first-line (84% ORR, 53% CR, estimated 2-year PFS rate, 84%; refs. 56, 57) settings.

Preclinical data suggest superiority of second-generation monoclonal antibodies versus rituximab, including GA-101 (obinutuzumab; ref. 79) and ofatumumab (80). Although ofatumumab showed modest activity in a recent phase II study (81), a dramatic response was reported in a patient with refractory, high leukemic phase MCL (82). ORR in the GAUGUIN phase II study (2 doses of obinutuzumab) in 21 heavily pretreated patients with diffuse large B-cell lymphoma (DLBCL) and MCL was 24% to 37%, with median DOR of 9.8 months (83). Additional ongoing studies are looking at second-generation monoclonal antibodies for newer immunochemotherapy regimens in MCL.

Several novel agents, including BH3 mimetic-type BCL2 inhibitors such as navitoclax (ABT-263; ref. 84) and obatoclax mesylate (85), showed modest activity; more recently, venetoclax (ABT-199; abstract results; ref. 86) showed very impressive phase I activity in MCL and is being tested in combination. PI3Kδ inhibitors (e.g., idelalisib) or other small molecules, including second-generation BTK inhibitors, are being developed (87). Adoptive transfer of autologous CAR T cells, genetically engineered to express anti-CD19 specificity demonstrated impressive results in NHL, including DLBCL and MCL, supporting planned studies focusing on MCL (77, 88). Other strategies to induce T-cell responses through checkpoint inhibitors (e.g., anti–PD-1 antibodies) show extremely promising results in a number of tumors and are currently being explored in different lymphomas subtypes (89, 90).

On the basis of evolving clinical options, our recommendations for sequencing therapies in MCL are outlined in Fig. 1. First-line treatment should be based on “functional general condition” (not just a “cutoff” of age 60 or 65 years), as well as a patient's ability to tolerate intensive therapy. Achieving an early CR in MCL very clearly impacts OS, and by extension, depth of CR. Molecular CR should be our goal moving forward and may also serve as an “endpoint” for treatment adjustment according to the interim molecular response. Dose-intense therapy with high-dose Ara-C–containing induction, with or without HDT-ASCT, translates into higher CR and molecular CR rates, with extended PFS (>5 years). The need to maintain HDT-ASCT after induction in patients with CR/molecular CR is currently debatable and is being evaluated in ongoing trials.

Figure 1.

Sequential treatment algorithm in patients with MCL: first-line treatment for younger, fit versus older and/or frail patients, consolidation or maintenance, and treatment for relapsed/refractory disease. First-line treatment is commonly selected on the basis of patient's age and condition, gauging a patient's tolerability to aggressive versus less aggressive induction immunochemotherapy. Patients eligible for HDT are typically considered for cytarabine-containing induction regimen, such as R-CHOP/R-DHAP followed by ASCT or R-hyper-CVAD if tolerated. Older, transplant-ineligible or unfit patients mainly receive BR, R-CHOP, or R-CHOP-bortezomib induction followed by rituximab maintenance. Non-myeloablative allogeneic transplant is a potential approach for special cases of fit patients who failed several lines of therapy; its use reflects a physician preference rather than significant clinical evidence. FDA-approved therapies for relapsed/refractory MCL include bortezomib, lenalidomide, and ibrutinib, whereas temsirolimus is approved in the EU. CVAD, cyclophosphamide, vincristine, doxorubicin, dexamethasone; EU, European Union; HD, high dose; PD, progressive disease; R, rituximab; R-CHOP, rituximab, cyclophosphamide, vincristine, doxorubicin, and prednisone; R-DHAP, rituximab, dexamethasone, cytarabine, cisplatin; SCT, stem cell transplantation.

Figure 1.

Sequential treatment algorithm in patients with MCL: first-line treatment for younger, fit versus older and/or frail patients, consolidation or maintenance, and treatment for relapsed/refractory disease. First-line treatment is commonly selected on the basis of patient's age and condition, gauging a patient's tolerability to aggressive versus less aggressive induction immunochemotherapy. Patients eligible for HDT are typically considered for cytarabine-containing induction regimen, such as R-CHOP/R-DHAP followed by ASCT or R-hyper-CVAD if tolerated. Older, transplant-ineligible or unfit patients mainly receive BR, R-CHOP, or R-CHOP-bortezomib induction followed by rituximab maintenance. Non-myeloablative allogeneic transplant is a potential approach for special cases of fit patients who failed several lines of therapy; its use reflects a physician preference rather than significant clinical evidence. FDA-approved therapies for relapsed/refractory MCL include bortezomib, lenalidomide, and ibrutinib, whereas temsirolimus is approved in the EU. CVAD, cyclophosphamide, vincristine, doxorubicin, dexamethasone; EU, European Union; HD, high dose; PD, progressive disease; R, rituximab; R-CHOP, rituximab, cyclophosphamide, vincristine, doxorubicin, and prednisone; R-DHAP, rituximab, dexamethasone, cytarabine, cisplatin; SCT, stem cell transplantation.

Close modal

In non–HDT-eligible patients, induction with R-chemotherapy and other biologicals such as VcR-CAP offers a new backbone for frontline FDA approval whereas BR is being tested as another platform for combinations. The use of maintenance is now supported in both younger and older patients, although the role and duration of maintenance beyond MRD-negative status is unclear, requiring further study.

In the relapsed/refractory setting, standard chemotherapy should not be the only option, considering its limited benefit and frequent chemoresistance. Novel therapies often provide durable responses but are not curative—only non-myeloablative transplantation may provide a cure. The development of novel combinations both in frontline and relapsed/refractory settings will likely expand and provide opportunities for non-chemotherapy options and/or chemotherapy combinations either sequentially or as maintenance/consolidation postinduction. Clinical trials should be the default in managing patients with MCL given the rapid changes in the field. Finally, efforts are needed to help translate the impressive biologic diversity of MCL into relevant clinical markers to better stratify patients and further improve the MCL treatment paradigm overall. MCL is changing, so let us continue to embrace those changes.

A. Goy reports receiving speakers bureau honoraria from and is a consultant/advisory board member for Celgene Corporation, Johnson & Johnson/Pharmacyclics, and Takeda. No potential conflicts of interest were disclosed by the other author.

The authors received editorial support in the preparation of the article from Julie Kern, PhD, CMPP, of Bio Connections LLC, which was funded by Celgene Corporation.

1.
Royo
C
,
Navarro
A
,
Clot
G
,
Salaverria
I
,
Gine
E
,
Jares
P
, et al
Non-nodal type of mantle cell lymphoma is a specific biological and clinical subgroup of the disease
.
Leukemia
2012
;
26
:
1895
8
.
2.
Goy
A
,
Kahl
B
. 
Mantle cell lymphoma: the promise of new treatment options
.
Crit Rev Oncol Hematol
2011
;
80
:
69
86
.
3.
Armitage
JO
,
Weisenburger
DD
. 
New approach to classifying non-Hodgkin's lymphomas: clinical features of the major histologic subtypes. Non-Hodgkin's Lymphoma Classification Project
.
J Clin Oncol
1998
;
16
:
2780
95
.
4.
Chandran
R
,
Gardiner
SK
,
Simon
M
,
Spurgeon
SE
. 
Survival trends in mantle cell lymphoma in the United States over 16 years 1992–2007
.
Leuk Lymphoma
2012
;
53
:
1488
93
.
5.
Perez-Galan
P
,
Dreyling
M
,
Wiestner
A
. 
Mantle cell lymphoma: biology, pathogenesis, and the molecular basis of treatment in the genomic era
.
Blood
2011
;
117
:
26
38
.
6.
Rosenwald
A
,
Wright
G
,
Wiestner
A
,
Chan
WC
,
Connors
JM
,
Campo
E
, et al
The proliferation gene expression signature is a quantitative integrator of oncogenic events that predicts survival in mantle cell lymphoma
.
Cancer Cell
2003
;
3
:
185
97
.
7.
Hoster
E
,
Dreyling
M
,
Klapper
W
,
Gisselbrecht
C
,
van Hoof
A
,
Kluin-Nelemans
HC
, et al
A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma
.
Blood
2008
;
111
:
558
65
.
8.
Hoster
E
,
Klapper
W
,
Hermine
O
,
Kluin-Nelemans
HC
,
Walewski
J
,
van Hoof
A
, et al
Confirmation of the mantle-cell lymphoma international prognostic index in randomized trials of the European mantle-cell lymphoma network
.
J Clin Oncol
2014
;
32
:
1338
46
.
9.
Abrahamsson
A
,
Albertsson-Lindblad
A
,
Brown
PN
,
Baumgartner-Wennerholm
S
,
Pedersen
LM
,
D'Amore
F
, et al
Real world data on primary treatment for mantle cell lymphoma: a Nordic Lymphoma Group observational study
.
Blood
2014
;
124
:
1288
95
.
10.
Leux
C
,
Maynadie
M
,
Troussard
X
,
Cabrera
Q
,
Herry
A
,
Le Guyader-Peyrou
S
, et al
Mantle cell lymphoma epidemiology: a population-based study in France
.
Ann Hematol
2014
;
93
:
1327
33
.
11.
Determann
O
,
Hoster
E
,
Ott
G
,
Wolfram Bernd
H
,
Loddenkemper
C
,
Leo Hansmann
M
, et al
Ki-67 predicts outcome in advanced-stage mantle cell lymphoma patients treated with anti-CD20 immunochemotherapy: results from randomized trials of the European MCL Network and the German Low Grade Lymphoma Study Group
.
Blood
2008
;
111
:
2385
7
.
12.
Husby
S
,
Ralfkiaer
U
,
Garde
C
,
Zandi
R
,
Ek
S
,
Kolstad
A
, et al
miR-18b overexpression identifies mantle cell lymphoma patients with poor outcome and improves the MIPI-B prognosticator
.
Blood
2015
;
125
:
2669
77
.
13.
Navarro
A
,
Clot
G
,
Royo
C
,
Jares
P
,
Hadzidimitriou
A
,
Agathangelidis
A
, et al
Molecular subsets of mantle cell lymphoma defined by the IGHV mutational status and SOX11 expression have distinct biologic and clinical features
.
Cancer Res
2012
;
72
:
5307
16
.
14.
Nordstrom
L
,
Sernbo
S
,
Eden
P
,
Gronbaek
K
,
Kolstad
A
,
Raty
R
, et al
SOX11 and TP53 add prognostic information to MIPI in a homogenously treated cohort of mantle cell lymphoma–a Nordic Lymphoma Group study
.
Br J Haematol
2014
;
166
:
98
108
.
15.
Bea
S
,
Valdes-Mas
R
,
Navarro
A
,
Salaverria
I
,
Martin-Garcia
D
,
Jares
P
, et al
Landscape of somatic mutations and clonal evolution in mantle cell lymphoma
.
Proc Natl Acad Sci U S A
2013
;
110
:
18250
5
.
16.
Martin
P
,
Chadburn
A
,
Christos
P
,
Weil
K
,
Furman
RR
,
Ruan
J
, et al
Outcome of deferred initial therapy in mantle-cell lymphoma
.
J Clin Oncol
2009
;
27
:
1209
13
.
17.
Eve
HE
,
Furtado
MV
,
Hamon
MD
,
Rule
SA
. 
Time to treatment does not influence overall survival in newly diagnosed mantle-cell lymphoma
.
J Clin Oncol
2009
;
27
:
e189
90
;
author reply e91
.
18.
Fernandez
V
,
Salamero
O
,
Espinet
B
,
Sole
F
,
Royo
C
,
Navarro
A
, et al
Genomic and gene expression profiling defines indolent forms of mantle cell lymphoma
.
Cancer Res
2010
;
70
:
1408
18
.
19.
Greiner
TC
,
Dasgupta
C
,
Ho
VV
,
Weisenburger
DD
,
Smith
LM
,
Lynch
JC
, et al
Mutation and genomic deletion status of ataxia telangiectasia mutated (ATM) and p53 confer specific gene expression profiles in mantle cell lymphoma
.
Proc Natl Acad Sci U S A
2006
;
103
:
2352
7
.
20.
Carvajal-Cuenca
A
,
Sua
LF
,
Silva
NM
,
Pittaluga
S
,
Royo
C
,
Song
JY
, et al
In situ mantle cell lymphoma: clinical implications of an incidental finding with indolent clinical behavior
.
Haematologica
2012
;
97
:
270
8
.
21.
Zelenetz
AD
,
Gordon
LI
,
Wierda
WG
,
Abramson
JS
,
Advani
RH
,
Andreadis
CB
, et al
Non-Hodgkin's lymphomas, version 4.2014
.
J Natl Compr Canc Netw
2014
;
12
:
1282
303
.
22.
Dreyling
M
,
Geisler
C
,
Hermine
O
,
Kluin-Nelemans
HC
,
Le Gouill
S
,
Rule
S
, et al
Newly diagnosed and relapsed mantle cell lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
2014
;
25
Suppl 3
:
iii83
92
.
23.
Dreyling
M
,
Lenz
G
,
Hoster
E
,
Van Hoof
A
,
Gisselbrecht
C
,
Schmits
R
, et al
Early consolidation by myeloablative radiochemotherapy followed by autologous stem cell transplantation in first remission significantly prolongs progression-free survival in mantle-cell lymphoma: results of a prospective randomized trial of the European MCL Network
.
Blood
2005
;
105
:
2677
84
.
24.
LaCasce
AS
,
Vandergrift
JL
,
Rodriguez
MA
,
Abel
GA
,
Crosby
AL
,
Czuczman
MS
, et al
Comparative outcome of initial therapy for younger patients with mantle cell lymphoma: an analysis from the NCCN NHL Database
.
Blood
2012
;
119
:
2093
9
.
25.
Delarue
R
,
Haioun
C
,
Ribrag
V
,
Brice
P
,
Delmer
A
,
Tilly
H
, et al
CHOP and DHAP plus rituximab followed by autologous stem cell transplantation in mantle cell lymphoma: a phase 2 study from the Groupe d'Etude des Lymphomes de l'Adulte
.
Blood
2013
;
121
:
48
53
.
26.
Romaguera
JE
,
Fayad
L
,
Rodriguez
MA
,
Broglio
KR
,
Hagemeister
FB
,
Pro
B
, et al
High rate of durable remissions after treatment of newly diagnosed aggressive mantle-cell lymphoma with rituximab plus hyper-CVAD alternating with rituximab plus high-dose methotrexate and cytarabine
.
J Clin Oncol
2005
;
23
:
7013
23
.
27.
Romaguera
JE
,
Fayad
LE
,
Feng
L
,
Hartig
K
,
Weaver
P
,
Rodriguez
MA
, et al
Ten-year follow-up after intense chemoimmunotherapy with Rituximab-HyperCVAD alternating with Rituximab-high dose methotrexate/cytarabine (R-MA) and without stem cell transplantation in patients with untreated aggressive mantle cell lymphoma
.
Br J Haematol
2010
;
150
:
200
8
.
28.
Geisler
CH
,
Kolstad
A
,
Laurell
A
,
Andersen
NS
,
Pedersen
LB
,
Jerkeman
M
, et al
Long-term progression-free survival of mantle cell lymphoma after intensive front-line immunochemotherapy with in vivo-purged stem cell rescue: a nonrandomized phase 2 multicenter study by the Nordic Lymphoma Group
.
Blood
2008
;
112
:
2687
93
.
29.
Geisler
CH
,
Kolstad
A
,
Laurell
A
,
Jerkeman
M
,
Raty
R
,
Andersen
NS
, et al
Nordic MCL2 trial update: six-year follow-up after intensive immunochemotherapy for untreated mantle cell lymphoma followed by BEAM or BEAC + autologous stem-cell support: still very long survival but late relapses do occur
.
Br J Haematol
2012
;
158
:
355
62
.
30.
Damon
LE
,
Johnson
JL
,
Niedzwiecki
D
,
Cheson
BD
,
Hurd
DD
,
Bartlett
NL
, et al
Immunochemotherapy and autologous stem-cell transplantation for untreated patients with mantle-cell lymphoma: CALGB 59909
.
J Clin Oncol
2009
;
27
:
6101
8
.
31.
Lossos
IS
,
Hosein
PJ
,
Morgensztern
D
,
Coleman
F
,
Escalon
MP
,
Byrne
GE
 Jr
, et al
High rate and prolonged duration of complete remissions induced by rituximab, methotrexate, doxorubicin, cyclophosphamide, vincristine, ifosfamide, etoposide, cytarabine, and thalidomide (R-MACLO-IVAM-T), a modification of the National Cancer Institute 89-C-41 regimen, in patients with newly diagnosed mantle cell lymphoma
.
Leuk Lymphoma
2010
;
51
:
406
14
.
32.
Hermine
O
,
Hoster
E
,
Walewski
J
,
Ribrag
V
,
Brousse
N
,
Thieblemont
C
, et al
Alternating courses of 3x CHOP and 3x DHAP plus rituximab followed by a high dose ARA-C containing myeloablative regimen and autologous stem cell transplantation (ASCT) is superior to 6 courses CHOP plus rituximab followed by myeloablative radiochemotherapy and ASCT in mantle cell lymphoma: results of the MCL Younger Trial of the European Mantle Cell Lymphoma Network (MCL net) [abstract]
. In: 
Proceedings of the 53rd ASH Annual Meeting and Exposition; 2011 Dec 10–13
;
Orlando, FL. Washington (DC)
:
American Society of Hematology
; 
2011
.
Abstract nr 110
.
33.
Hermine
O
,
Hoster
E
,
Walewski
J
,
Ribrag
V
,
Brousse
N
,
Thieblemont
C
, et al
Alternating courses of 3x CHOP and 3x DHAP plus rituximab followed by a high dose ARA-C containing myeloablative regimen and autologous stem cell transplantation (ASCT) increases overall survival when compared to 6 courses of CHOP plus rituximab followed by myeloablative radiochemotherapy and ASCT in mantle cell lymphoma: final analysis of the MCL Younger Trial of the European Mantle Cell Lymphoma Network (MCL net) [abstract]
. In: 
Proceedings of the 54th ASH Annual Meeting and Exposition; 2012 Dec 9–11
;
Atlanta, GA. Washington (DC)
:
American Society of Hematology
; 
2012
.
Abstract nr 151
.
34.
Pott
C
,
Hoster
E
,
Delfau-Larue
M-H
,
Beldjord
K
,
Böttcher
S
,
Asnafi
V
, et al
Molecular remission is an independent predictor of clinical outcome in patients with mantle cell lymphoma after combined immunochemotherapy: a European MCL intergroup study
.
Blood
2010
;
115
:
3215
23
.
35.
Touzeau
C
,
Leux
C
,
Bouabdallah
R
,
Roussel
M
,
Delarue
R
,
Bouabdallah
K
, et al
Autologous stem cell transplantation in mantle cell lymphoma: a report from the SFGM-TC
.
Ann Hematol
2014
;
93
:
233
42
.
36.
Le Gouill
S
,
Thieblemont
C
,
Oberic
L
,
Bouabdallah
K
,
Gyan
E
,
Damaj
G
, et al
Rituximab maintenance versus wait and watch after four courses of R-DHAP followed by autologous stem cell transplantation in previously untreated young patients with mantle cell lymphoma: first interim analysis of the phase III prospective Lyma trial, a Lysa study [abstract]
. In: 
Proceedings of the 56th ASH Annual Meeting and Exposition; 2014 Dec 6–9
;
San Francisco, CA. Washington (DC)
:
American Society of Hematology
; 
2014
.
Abstract nr 146
.
37.
Howard
OM
,
Gribben
JG
,
Neuberg
DS
,
Grossbard
M
,
Poor
C
,
Janicek
MJ
, et al
Rituximab and CHOP induction therapy for newly diagnosed mantle-cell lymphoma: molecular complete responses are not predictive of progression-free survival
.
J Clin Oncol
2002
;
20
:
1288
94
.
38.
Lenz
G
,
Dreyling
M
,
Hoster
E
,
Wormann
B
,
Duhrsen
U
,
Metzner
B
, et al
Immunochemotherapy with rituximab and cyclophosphamide, doxorubicin, vincristine, and prednisone significantly improves response and time to treatment failure, but not long-term outcome in patients with previously untreated mantle cell lymphoma: results of a prospective randomized trial of the German Low Grade Lymphoma Study Group (GLSG)
.
J Clin Oncol
2005
;
23
:
1984
92
.
39.
Kluin-Nelemans
HC
,
Hoster
E
,
Hermine
O
,
Walewski
J
,
Trneny
M
,
Geisler
CH
, et al
Treatment of older patients with mantle-cell lymphoma
.
N Engl J Med
2012
;
367
:
520
31
.
40.
Raty
R
,
Honkanen
T
,
Jantunen
E
,
Jyrkkio
S
,
Karjalainen-Lindsberg
ML
,
Kuittinen
O
, et al
Prolonged immunochemotherapy with rituximab, cytarabine and fludarabine added to cyclophosphamide, doxorubicin, vincristine and prednisolone and followed by rituximab maintenance in untreated elderly patients with mantle cell lymphoma: a prospective study by the Finnish Lymphoma Group
.
Leuk Lymphoma
2012
;
53
:
1920
8
.
41.
Ruan
J
,
Martin
P
,
Furman
RR
,
Lee
SM
,
Cheung
K
,
Vose
JM
, et al
Bortezomib plus CHOP-rituximab for previously untreated diffuse large B-cell lymphoma and mantle cell lymphoma
.
J Clin Oncol
2011
;
29
:
690
7
.
42.
Robak
T
,
Huang
H
,
Jin
J
,
Zhu
J
,
Liu
T
,
Samoilova
O
, et al
Bortezomib-based therapy for newly diagnosed mantle-cell lymphoma
.
N Engl J Med
2015
;
372
:
944
53
.
43.
Velcade (bortezomib) for injection prescribing information [PDF on the Internet]
.
Cambridge, MA
:
Millennium Pharmaceuticals
; 
2014
[cited 2015 Jan 14]. Available from
: http://www.velcade.com/Files/PDFs/VELCADE_PRESCRIBING_INFORMATION.pdf.
44.
Bernstein
SH
,
Epner
E
,
Unger
JM
,
Leblanc
M
,
Cebula
E
,
Burack
R
, et al
A phase II multicenter trial of hyperCVAD MTX/Ara-C and rituximab in patients with previously untreated mantle cell lymphoma; SWOG 0213
.
Ann Oncol
2013
;
24
:
1587
93
.
45.
Chang
JE
,
Peterson
C
,
Choi
S
,
Eickhoff
JC
,
Kim
K
,
Yang
DT
, et al
VcR-CVAD induction chemotherapy followed by maintenance rituximab in mantle cell lymphoma: a Wisconsin Oncology Network study
.
Br J Haematol
2011
;
155
:
190
7
.
46.
Chang
JE
,
Li
H
,
Smith
MR
,
Gascoyne
RD
,
Paietta
EM
,
Yang
DT
, et al
Phase 2 study of VcR-CVAD with maintenance rituximab for untreated mantle cell lymphoma: an Eastern Cooperative Oncology Group study (E1405)
.
Blood
2014
;
123
:
1665
73
.
47.
Rummel
MJ
,
Niederle
N
,
Maschmeyer
G
,
Banat
GA
,
von Grunhagen
U
,
Losem
C
, et al
Bendamustine plus rituximab versus CHOP plus rituximab as first-line treatment for patients with indolent and mantle-cell lymphomas: an open-label, multicentre, randomised, phase 3 non-inferiority trial
.
Lancet
2013
;
381
:
1203
10
.
48.
Flinn
IW
,
van der Jagt
R
,
Kahl
BS
,
Wood
P
,
Hawkins
TE
,
Macdonald
D
, et al
Randomized trial of bendamustine-rituximab or R-CHOP/R-CVP in first-line treatment of indolent NHL or MCL: the BRIGHT study
.
Blood
2014
;
123
:
2944
52
.
49.
Friedberg
JW
,
Vose
JM
,
Kelly
JL
,
Young
F
,
Bernstein
SH
,
Peterson
D
, et al
The combination of bendamustine, bortezomib, and rituximab for patients with relapsed/refractory indolent and mantle cell non-Hodgkin lymphoma
.
Blood
2011
;
117
:
2807
12
.
50.
Gressin
R
,
Callanan
M
,
Daguindau
N
,
Tempescul
A
,
Carras
S
,
Moles
MP
, et al
Frontline therapy with the Ribvd regimen elicits high clinical and molecular response rates and long PFS in elderly patients mantle cell lymphoma: final results of a prospective phase II trial by the Lysa group [abstract]
. In: 
Proceedings of the 56th ASH Annual Meeting and Exposiyion; 2014 Dec 6–9
;
San Francisco, CA. Washington (DC)
:
American Society for Hematology
; 
2014
.
Abstract nr 148
.
51.
Visco
C
,
Finotto
S
,
Zambello
R
,
Paolini
R
,
Menin
A
,
Zanotti
R
, et al
Combination of rituximab, bendamustine, and cytarabine for patients with mantle-cell non-Hodgkin lymphoma ineligible for intensive regimens or autologous transplantation
.
J Clin Oncol
2013
;
31
:
1442
9
.
52.
Till
BG
,
Li
H
,
Bernstein
SH
,
Fisher
RI
,
Burack
R
,
Rimsza
LM
, et al
Phase II trial of R-CHOP pus bortezomib induction therapy followed by bortezomib maintenance for previously untreated mantle cell lymphoma: SWOG 0601 [abstract]
. In: 
Proceedings of the 56th ASH Annual Meeting and Exposition; 2014 Dec 6–9
;
San Francisco, CA. Washington (DC)
:
American Society of Hematology
; 
2014
.
Abstract nr 149
.
53.
Furtado
M
,
Johnson
R
,
Kruger
A
,
Turner
D
,
Rule
S
. 
Addition of bortezomib to standard dose chop chemotherapy improves response and survival in relapsed mantle cell lymphoma
.
Br J Haematol
2015
;
168
:
55
62
.
54.
Kaplan
LD
,
Jung
S
,
Bartelet
N
,
Johnson
J
,
Byrd
JC
,
Blum
KA
, et al
Bortezomib maintenance (BM) versus consolidation (BC) following aggressive immunochemotherapy and autologous stem cell transplant (ASCT) for untreated mantle cell lymphoma (MCL): CALGB 50403 [abstract]
. In: 
Proceedings of the 12th International Conference in Malignant Lymphoma; 2013 Jun 19–22
;
Lugano, Switzerland. Bellinzona (Switzerland)
:
Istituto Oncologico della Svizzera Italiana (IOSI)
; 
2013
.
Abstract nr 089
.
55.
Goy
A
,
Sinha
R
,
Williams
ME
,
Kalayoglu Besisik
S
,
Drach
J
,
Ramchandren
R
, et al
Single-agent lenalidomide in patients with mantle-cell lymphoma who relapsed or progressed after or were refractory to bortezomib: phase II MCL-001 (EMERGE) study
.
J Clin Oncol
2013
;
31
:
3688
95
.
56.
Wang
M
,
Fayad
L
,
Wagner-Bartak
N
,
Zhang
L
,
Hagemeister
F
,
Neelapu
SS
, et al
Lenalidomide in combination with rituximab for patients with relapsed or refractory mantle-cell lymphoma: a phase 1/2 clinical trial
.
Lancet Oncol
2012
;
13
:
716
23
.
57.
Ruan
J
,
Martin
P
,
Shah
BD
,
Schuster
SJ
,
Smith
SM
,
Furman
RR
, et al
Sustained remission with the combination biologic doublet of lenalidomide plus rituximab as initial treatment for mantle cell lymphoma: a multicenter phase II study report [abstract]
. In: 
Proceedings of the 56th ASH Annual Meeting and Exposition; 2014 Dec 6–9; San Francisco, CA
.
Washington (DC)
:
American Society of Hematology
; 
2014
.
Abstract nr 625
.
58.
Smith
MR
,
Li
H
,
Gordon
L
,
Gascoyne
RD
,
Paietta
E
,
Forero-Torres
A
, et al
Phase II study of rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone immunochemotherapy followed by yttrium-90-ibritumomab tiuxetan in untreated mantle-cell lymphoma: Eastern Cooperative Oncology Group Study E1499
.
J Clin Oncol
2012
;
30
:
3119
26
.
59.
Kolstad
A
,
Laurell
A
,
Jerkeman
M
,
Gronbaek
K
,
Elonen
E
,
Raty
R
, et al
Nordic MCL3 study: 90Y-ibritumomab-tiuxetan added to BEAM/C in non-CR patients before transplant in mantle cell lymphoma
.
Blood
2014
;
123
:
2953
9
.
60.
Mato
AR
,
Svoboda
J
,
Feldman
T
,
Zielonka
T
,
Agress
H
,
Panush
D
, et al
Post-treatment (not interim) positron emission tomography-computed tomography scan status is highly predictive of outcome in mantle cell lymphoma patients treated with R-HyperCVAD
.
Cancer
2012
;
118
:
3565
70
.
61.
Cohen
JB
,
Hall
NC
,
Ruppert
AS
,
Jones
JA
,
Porcu
P
,
Baiocchi
R
, et al
Association of pre-transplantation positron emission tomography/computed tomography and outcome in mantle cell lymphoma
.
Bone Marrow Transplant
2013
;
48
:
1212
7
.
62.
Kedmi
M
,
Avivi
I
,
Ribakovsky
E
,
Benyamini
N
,
Davidson
T
,
Goshen
E
, et al
Is there a role for therapy response assessment with FDG-PET/CT in mantle cell lymphoma?
Leuk Lymphoma
2014
;
55
:
2484
9
.
63.
Liu
H
,
Johnson
JL
,
Koval
G
,
Malnassy
G
,
Sher
D
,
Damon
LE
, et al
Detection of minimal residual disease following induction immunochemotherapy predicts progression free survival in mantle cell lymphoma: final results of CALGB 59909
.
Haematologica
2012
;
97
:
579
85
.
64.
Pott
C
,
Macintyre
E
,
Delfau-Larue
MH
,
Ribrag
V
,
Unterhalt
M
,
Kneba
M
, et al
MRD eradication should be the therapeutic goal in mantle cell lymphoma and may enable tailored treatment approaches: results of the intergroup trials of the European MCL Network [abstract]
. In: 
Proceedings of the 56th ASH Annual Meeting and Exposition; 2014 Dec 6–9
;
San Francisco, CA. Washington (DC)
:
American Society of Hematology
; 
2014
.
Abstract nr 147
.
65.
Rummel
MJ
,
Al-Batran
SE
,
Kim
SZ
,
Welslau
M
,
Hecker
R
,
Kofahl-Krause
D
, et al
Bendamustine plus rituximab is effective and has a favorable toxicity profile in the treatment of mantle cell and low-grade non-Hodgkin's lymphoma
.
J Clin Oncol
2005
;
23
:
3383
9
.
66.
Fisher
RI
,
Bernstein
SH
,
Kahl
BS
,
Djulbegovic
B
,
Robertson
MJ
,
de Vos
S
, et al
Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma
.
J Clin Oncol
2006
;
24
:
4867
74
.
67.
Goy
A
,
Bernstein
SH
,
Kahl
BS
,
Djulbegovic
B
,
Robertson
MJ
,
de Vos
S
, et al
Bortezomib in patients with relapsed or refractory mantle cell lymphoma: updated time-to-event analyses of the multicenter phase 2 PINNACLE study
.
Ann Oncol
2009
;
20
:
520
5
.
68.
Hess
G
,
Herbrecht
R
,
Romaguera
J
,
Verhoef
G
,
Crump
M
,
Gisselbrecht
C
, et al
Phase III study to evaluate temsirolimus compared with investigator's choice therapy for the treatment of relapsed or refractory mantle cell lymphoma
.
J Clin Oncol
2009
;
27
:
3822
9
.
69.
Wang
ML
,
Rule
S
,
Martin
P
,
Goy
A
,
Auer
R
,
Kahl
BS
, et al
Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma
.
N Engl J Med
2013
;
369
:
507
16
.
70.
Habermann
TM
,
Lossos
IS
,
Justice
G
,
Vose
JM
,
Wiernik
PH
,
McBride
K
, et al
Lenalidomide oral monotherapy produces a high response rate in patients with relapsed or refractory mantle cell lymphoma
.
Br J Haematol
2009
;
145
:
344
9
.
71.
Zinzani
PL
,
Vose
JM
,
Czuczman
MS
,
Reeder
CB
,
Haioun
C
,
Polikoff
J
, et al
Long-term follow-up of lenalidomide in relapsed/refractory mantle cell lymphoma: subset analysis of the NHL-003 study
.
Ann Oncol
2013
;
24
:
2892
7
.
72.
Trneny
M
,
Lamy
T
,
Walewski
J
,
Jurczak
W
,
Belada
D
,
Mayer
J
, et al
Phase II randomized, multicenter study of lenalidomide vs best investigator's choice in relapsed/refractory mantle cell lymphoma: results of the MCL-002 (SPRINT) study [abstract]
. In: 
Proceedings of the 56th ASH Annual Meeting and Exposition; 2014 Dec 6–9
;
San Francisco, CA. Washington (DC)
:
American Society of Hematology
; 
2014
.
Abstract nr 626
.
73.
Dietrich
S
,
Boumendil
A
,
Finel
H
,
Avivi
I
,
Volin
L
,
Cornelissen
J
, et al
Outcome and prognostic factors in patients with mantle-cell lymphoma relapsing after autologous stem-cell transplantation: a retrospective study of the European Group for Blood and Marrow Transplantation (EBMT)
.
Ann Oncol
2014
;
25
:
1053
8
.
74.
Robinson
S
,
Dreger
P
,
Caballero
D
,
Corradini
P
,
Geisler
C
,
Ghielmini
M
, et al
The EBMT/EMCL consensus project on the role of autologous and allogeneic stem cell transplantation in mantle cell lymphoma
.
Leukemia
2015
;
29
:
464
73
.
75.
Hamadani
M
,
Saber
W
,
Ahn
KW
,
Carreras
J
,
Cairo
MS
,
Fenske
TS
, et al
Allogeneic hematopoietic cell transplantation for chemotherapy-unresponsive mantle cell lymphoma: a cohort analysis from the center for international blood and marrow transplant research
.
Biol Blood Marrow Transplant
2013
;
19
:
625
31
.
76.
Tam
CS
,
Bassett
R
,
Ledesma
C
,
Korbling
M
,
Alousi
A
,
Hosing
C
, et al
Mature results of the M. D. Anderson Cancer Center risk-adapted transplantation strategy in mantle cell lymphoma
.
Blood
2009
;
113
:
4144
52
.
77.
Kochenderfer
JN
,
Dudley
ME
,
Kassim
SH
,
Somerville
RP
,
Carpenter
RO
,
Stetler-Stevenson
M
, et al
Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor
.
J Clin Oncol
2015
;
33
:
540
9
.
78.
Kaufmann
H
,
Raderer
M
,
Wohrer
S
,
Puspok
A
,
Bankier
A
,
Zielinski
C
, et al
Antitumor activity of rituximab plus thalidomide in patients with relapsed/refractory mantle cell lymphoma
.
Blood
2004
;
104
:
2269
71
.
79.
Mossner
E
,
Brunker
P
,
Moser
S
,
Puntener
U
,
Schmidt
C
,
Herter
S
, et al
Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity
.
Blood
2010
;
115
:
4393
402
.
80.
Castillo
J
,
Milani
C
,
Mendez-Allwood
D
. 
Ofatumumab, a second-generation anti-CD20 monoclonal antibody, for the treatment of lymphoproliferative and autoimmune disorders
.
Expert Opin Investig Drugs
2009
;
18
:
491
500
.
81.
Furtado
M
,
Dyer
MJ
,
Johnson
R
,
Berrow
M
,
Rule
S
. 
Ofatumumab monotherapy in relapsed/refractory mantle cell lymphoma–a phase II trial
.
Br J Haematol
2014
;
165
:
575
8
.
82.
Hunstig
F
,
Hammersen
J
,
Kunert
C
,
Petersen
I
,
Merz
H
,
Glaser
A
, et al
Complete remission after treatment with single-agent ofatumumab in a patient with high-risk leukemic mantle-cell lymphoma
.
J Clin Oncol
2013
;
31
:
e312
5
.
83.
Morschhauser
FA
,
Cartron
G
,
Thieblemont
C
,
Solal-Celigny
P
,
Haioun
C
,
Bouabdallah
R
, et al
Obinutuzumab (GA101) monotherapy in relapsed/refractory diffuse large b-cell lymphoma or mantle-cell lymphoma: results from the phase II GAUGUIN study
.
J Clin Oncol
2013
;
31
:
2912
9
.
84.
Wilson
WH
,
O'Connor
OA
,
Czuczman
MS
,
LaCasce
AS
,
Gerecitano
JF
,
Leonard
JP
, et al
Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity
.
Lancet Oncol
2010
;
11
:
1149
59
.
85.
Goy
A
,
Hernandez-Ilzaliturri
FJ
,
Kahl
B
,
Ford
P
,
Protomastro
E
,
Berger
M
. 
A phase I/II study of the pan Bcl-2 inhibitor obatoclax mesylate plus bortezomib for relapsed or refractory mantle cell lymphoma
.
Leuk Lymphoma
2014
;
55
:
2761
8
.
86.
Davids
MS
,
Seymour
JF
,
Gerecitano
JF
,
Kahl
BS
,
Pagel
JM
,
Wierda
WG
, et al
Phase I study of ABT-199 (GDC-0199) in patients with relapsed/refractory non-Hodgkin lymphoma: Responses observed in diffuse large B-cell and follicular lymphoma at higher cohort doses
.
J Clin Oncol
32
:
5s
, 
2014
(
suppl; abstr 8522
).
87.
Kahl
BS
,
Spurgeon
SE
,
Furman
RR
,
Flinn
IW
,
Coutre
SE
,
Brown
JR
, et al
A phase 1 study of the PI3Kdelta inhibitor idelalisib in patients with relapsed/refractory mantle cell lymphoma (MCL)
.
Blood
2014
;
123
:
3398
405
.
88.
Turtle
CJ
,
Sommermeyer
D
,
Berger
C
,
Hudecek
M
,
Shank
DM
,
Steevens
NN
, et al
Therapy of B cell malignancies with CD19-specific chimeric antigen receptor-modified T cells of defined subset composition [abstract]
. In: 
Proceedings of the 56th ASH Annual Meeting and Exposition; 2014 Dec 6–9
;
San Francisco, CA. Washington (DC)
:
American Society of Hematology
; 
2014
.
Abstract nr 384
.
89.
Olive
D
,
Le
KS
,
Roussel
M
,
Broussais
F
,
Azzaoui
I
,
Rossille
D
, et al
Diffuse large B cell lymphoma (DLBCL) infiltrating T cells display an activated and exhausted status and are inhibited by ligands of cosignaling receptors including PD-L1, PD-L2 and CD80 expressed by most DLBCL in situ [abstract]
. In: 
Proceedings of the 56th ASH Annual Meeting and Exposition; 2014 Dec 6–9
;
San Francisco, CA. Washington (DC)
:
American Society of Hematology
; 
2014
.
Abstract nr 1665
.
90.
Westin
JR
,
Chu
F
,
Zhang
M
,
Fayad
LE
,
Kwak
LW
,
Fowler
N
, et al
Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial
.
Lancet Oncol
2014
;
15
:
69
77
.